Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.634
Filtrar
1.
J Pharmacol Sci ; 155(2): 21-28, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38677782

RESUMEN

Goblet cell hyperplasia and increased mucus production are features of airway diseases, including asthma, and excess airway mucus often worsens these conditions. Even steroids are not uniformly effective in mucus production in severe asthma, and new therapeutic options are needed. Seihaito is a Japanese traditional medicine that is used clinically as an antitussive and expectorant. In the present study, we examined the effect of Seihaito on goblet cell differentiation and mucus production. In in vitro studies, using air-liquid interface culture of guinea-pig tracheal epithelial cells, Seihaito inhibited IL-13-induced proliferation of goblet cells and MUC5AC, a major component of mucus production. Seihaito suppressed goblet cell-specific gene expression, without changing ciliary cell-specific genes, suggesting that it inhibits goblet cell differentiation. In addition, Seihaito suppressed MUC5AC expression in cells transfected with SPDEF, a transcription factor activated by IL-13. Furthermore, Seihaito attenuated in vivo goblet cell proliferation and MUC5AC mRNA expression in IL-13-treated mouse lungs. Collectively, these findings demonstrated that Seihaito has an inhibitory effect on goblet cell differentiation and mucus production, which is at least partly due to the inhibition of SPDEF.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Células Caliciformes , Interleucina-13 , Medicina Kampo , Metaplasia , Mucina 5AC , Moco , Animales , Células Caliciformes/efectos de los fármacos , Células Caliciformes/patología , Células Caliciformes/metabolismo , Interleucina-13/metabolismo , Mucina 5AC/genética , Mucina 5AC/metabolismo , Moco/metabolismo , Diferenciación Celular/efectos de los fármacos , Cobayas , Proliferación Celular/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Células Cultivadas , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , Masculino , Expresión Génica/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Ratones , Tráquea/citología , Tráquea/efectos de los fármacos , Tráquea/patología , Tráquea/metabolismo
2.
Science ; 384(6693): 295-301, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38669574

RESUMEN

Airway neuroendocrine (NE) cells have been proposed to serve as specialized sensory epithelial cells that modulate respiratory behavior by communicating with nearby nerve endings. However, their functional properties and physiological roles in the healthy lung, trachea, and larynx remain largely unknown. In this work, we show that murine NE cells in these compartments have distinct biophysical properties but share sensitivity to two commonly aspirated noxious stimuli, water and acid. Moreover, we found that tracheal and laryngeal NE cells protect the airways by releasing adenosine 5'-triphosphate (ATP) to activate purinoreceptive sensory neurons that initiate swallowing and expiratory reflexes. Our work uncovers the broad molecular and biophysical diversity of NE cells across the airways and reveals mechanisms by which these specialized excitable cells serve as sentinels for activating protective responses.


Asunto(s)
Adenosina Trifosfato , Laringe , Células Neuroendocrinas , Reflejo , Tráquea , Animales , Ratones , Células Neuroendocrinas/metabolismo , Laringe/fisiología , Adenosina Trifosfato/metabolismo , Reflejo/fisiología , Tráquea/inervación , Tráquea/citología , Deglución , Pulmón/fisiología , Espiración/fisiología , Agua/metabolismo , Células Receptoras Sensoriales/fisiología , Ratones Endogámicos C57BL
3.
Science ; 384(6693): 269-270, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38669581
4.
Nature ; 621(7980): 857-867, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37730992

RESUMEN

Speciation leads to adaptive changes in organ cellular physiology and creates challenges for studying rare cell-type functions that diverge between humans and mice. Rare cystic fibrosis transmembrane conductance regulator (CFTR)-rich pulmonary ionocytes exist throughout the cartilaginous airways of humans1,2, but limited presence and divergent biology in the proximal trachea of mice has prevented the use of traditional transgenic models to elucidate ionocyte functions in the airway. Here we describe the creation and use of conditional genetic ferret models to dissect pulmonary ionocyte biology and function by enabling ionocyte lineage tracing (FOXI1-CreERT2::ROSA-TG), ionocyte ablation (FOXI1-KO) and ionocyte-specific deletion of CFTR (FOXI1-CreERT2::CFTRL/L). By comparing these models with cystic fibrosis ferrets3,4, we demonstrate that ionocytes control airway surface liquid absorption, secretion, pH and mucus viscosity-leading to reduced airway surface liquid volume and impaired mucociliary clearance in cystic fibrosis, FOXI1-KO and FOXI1-CreERT2::CFTRL/L ferrets. These processes are regulated by CFTR-dependent ionocyte transport of Cl- and HCO3-. Single-cell transcriptomics and in vivo lineage tracing revealed three subtypes of pulmonary ionocytes and a FOXI1-lineage common rare cell progenitor for ionocytes, tuft cells and neuroendocrine cells during airway development. Thus, rare pulmonary ionocytes perform critical CFTR-dependent functions in the proximal airway that are hallmark features of cystic fibrosis airway disease. These studies provide a road map for using conditional genetics in the first non-rodent mammal to address gene function, cell biology and disease processes that have greater evolutionary conservation between humans and ferrets.


Asunto(s)
Fibrosis Quística , Modelos Animales de Enfermedad , Hurones , Pulmón , Transgenes , Animales , Humanos , Animales Modificados Genéticamente , Linaje de la Célula , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Hurones/genética , Hurones/fisiología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Pulmón/citología , Pulmón/metabolismo , Pulmón/patología , Tráquea/citología , Transgenes/genética
5.
Development ; 149(3)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35112129

RESUMEN

The tracheal epithelium is a primary target for pulmonary diseases as it provides a conduit for air flow between the environment and the lung lobes. The cellular and molecular mechanisms underlying airway epithelial cell proliferation and differentiation remain poorly understood. Hedgehog (HH) signaling orchestrates communication between epithelial and mesenchymal cells in the lung, where it modulates stromal cell proliferation, differentiation and signaling back to the epithelium. Here, we reveal a previously unreported autocrine function of HH signaling in airway epithelial cells. Epithelial cell depletion of the ligand sonic hedgehog (SHH) or its effector smoothened (SMO) causes defects in both epithelial cell proliferation and differentiation. In cultured primary human airway epithelial cells, HH signaling inhibition also hampers cell proliferation and differentiation. Epithelial HH function is mediated, at least in part, through transcriptional activation, as HH signaling inhibition leads to downregulation of cell type-specific transcription factor genes in both the mouse trachea and human airway epithelial cells. These results provide new insights into the role of HH signaling in epithelial cell proliferation and differentiation during airway development.


Asunto(s)
Comunicación Autocrina/fisiología , Diferenciación Celular , Proliferación Celular , Proteínas Hedgehog/metabolismo , Transducción de Señal/genética , Animales , Células Cultivadas , Regulación hacia Abajo , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Proteínas Hedgehog/deficiencia , Proteínas Hedgehog/genética , Humanos , Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Noqueados , Receptor Smoothened/deficiencia , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Tráquea/citología , Tráquea/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Int J Mol Sci ; 23(3)2022 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-35162999

RESUMEN

Influenza (IAV) neuraminidase (NA) is a glycoprotein required for the viral exit from the cell. NA requires disulfide bonds for proper function. We have recently demonstrated that protein disulfide isomerase (PDI)A3 is required for oxidative folding of IAV hemagglutinin (HA), and viral propagation. However, it not known whether PDIs are required for NA maturation or if these interactions represent a putative target for the treatment of influenza infection. We sought to determine whether PDIA3 is required for disulfide bonds of NA, its activity, and propagation of the virus. Requirement of disulfides for NA oligomerization and activity were determined using biotin switch and redox assays in WT and PDIA3-/- in A549 cells. A PDI specific inhibitor (LOC14) was utilized to determine the requirement of PDIs in NA activity, IAV burden, and inflammatory response in A549 and primary mouse tracheal epithelial cells. Mice were treated with the inhibitor LOC14 and subsequently examined for IAV burden, NA activity, cytokine, and immune response. IAV-NA interacts with PDIA3 and this interaction is required for NA activity. PDIA3 ablation or inhibition decreased NA activity, viral burden, and inflammatory response in lung epithelial cells. LOC14 treatment significantly attenuated the influenza-induced inflammatory response in mice including the overall viral burden. These results provide evidence for PDIA3 inhibition suppressing NA activity, potentially providing a novel platform for host-targeted antiviral therapies.


Asunto(s)
Inhibidores Enzimáticos/administración & dosificación , Subtipo H1N1 del Virus de la Influenza A/enzimología , Neuraminidasa/metabolismo , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Proteína Disulfuro Isomerasas/metabolismo , Proteínas Virales/metabolismo , Células A549 , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Perros , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Células de Riñón Canino Madin Darby , Ratones , Neuraminidasa/química , Infecciones por Orthomyxoviridae/metabolismo , Cultivo Primario de Células , Pliegue de Proteína , Tráquea/citología , Tráquea/efectos de los fármacos , Tráquea/metabolismo , Tráquea/virología , Proteínas Virales/química
7.
Methods Mol Biol ; 2436: 157-165, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33950378

RESUMEN

Long-segment airway stenosis as well as their neoplastic transformation is life-threatening and still currently represent unsolved clinical problems. Indeed, despite several attempts, definitive surgical procedures are not presently available, and a suitable tracheal reconstruction or replacement remains an urgent clinical need. A possible innovative strategic solution to restore upper airway function may be represented by the creation of a bioprosthetic trachea, obtained through the combination of tissue engineering and regenerative medicine.Here we describe a two-step protocol for the ex vivo generation of tracheal segments. The first step involves the application of a decellularization technique that allows for the production of a naturally derived extracellular matrix (ECM)-based bio-scaffold, that maintains the macro- and micro-architecture as well as 9 the matrix-related signals distinctive of the original tissue. In the second step chondrocytes are seeded onto decellularized trachea, using a rotating bioreactor to ensure a correct scaffold repopulation.This multi-step approach represents a powerful tool for in vitro reconstruction of a bioengineered trachea that may constitute a promising solution to restore upper airway function. In addition, the procedures here described allow for the creation of a suitable 3D platform that may find useful applications, both for toxicological studies as well as organ transplantation strategies.


Asunto(s)
Condrocitos , Ingeniería de Tejidos , Andamios del Tejido , Tráquea , Reactores Biológicos , Condrocitos/citología , Prótesis e Implantes , Diseño de Prótesis , Ingeniería de Tejidos/métodos , Tráquea/citología , Tráquea/cirugía
8.
J Photochem Photobiol B ; 226: 112357, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34798503

RESUMEN

Mitochondrial antiviral signaling (MAVS) protein mediates innate antiviral responses, including responses to certain coronaviruses such as severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). We have previously shown that ultraviolet-A (UVA) therapy can prevent virus-induced cell death in human ciliated tracheal epithelial cells (HTEpC) infected with coronavirus-229E (CoV-229E), and results in increased intracellular levels of MAVS. In this study, we explored the mechanisms by which UVA light can activate MAVS, and whether local UVA light application can activate MAVS at locations distant from the light source (e.g. via cell-to-cell communication). MAVS levels were compared in HTEpC exposed to 2 mW/cm2 narrow band (NB)-UVA for 20 min and in unexposed controls at 30-40% and at 100% confluency, and in unexposed HTEpC treated with supernatants or lysates from UVA-exposed cells or from unexposed controls. MAVS was also assessed in different sections of confluent monolayer plates where only one section was exposed to NB-UVA. Our results showed that UVA increases the expression of MAVS protein. Further, cells in a confluent monolayer exposed to UVA conferred an elevation in MAVS in cells adjacent to the exposed section, and also in cells in the most distant sections which were not exposed to UVA. In this study, human ciliated tracheal epithelial cells exposed to UVA demonstrate increased MAVS protein, and also appear to transmit this influence to confluent cells not exposed to UVA, likely via cell-cell signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/efectos de la radiación , Rayos Ultravioleta , Proteínas Adaptadoras Transductoras de Señales/inmunología , COVID-19/inmunología , COVID-19/radioterapia , COVID-19/virología , Comunicación Celular/inmunología , Comunicación Celular/efectos de la radiación , Células Cultivadas , Células Epiteliales/inmunología , Células Epiteliales/efectos de la radiación , Interacciones Microbiota-Huesped/inmunología , Interacciones Microbiota-Huesped/efectos de la radiación , Humanos , Inmunidad Innata/efectos de la radiación , Fotobiología , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Transducción de Señal/inmunología , Transducción de Señal/efectos de la radiación , Tráquea/citología , Terapia Ultravioleta
9.
J Cell Mol Med ; 25(23): 10869-10878, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34725901

RESUMEN

Tissue engineering (TE) of long tracheal segments is conceptually appealing for patients with inoperable tracheal pathology. In tracheal TE, stem cells isolated from bone marrow or adipose tissue have been employed, but the ideal cell source has yet to be determined. When considering the origin of stem cells, cells isolated from a source embryonically related to the trachea may be more similar. In this study, we investigated the feasibility of isolating progenitor cells from pleura and pericard as an alternative cells source for tracheal tissue engineering. Porcine progenitor cells were isolated from pleura, pericard, trachea and adipose tissue and expanded in culture. Isolated cells were characterized by PCR, RNA sequencing, differentiation assays and cell survival assays and were compared to trachea and adipose-derived progenitor cells. Progenitor-like cells were successfully isolated and expanded from pericard and pleura as indicated by gene expression and functional analyses. Gene expression analysis and RNA sequencing showed a stem cell signature indicating multipotency, albeit that subtle differences between different cell sources were visible. Functional analysis revealed that these cells were able to differentiate towards chondrogenic, osteogenic and adipogenic lineages. Isolation of progenitor cells from pericard and pleura with stem cell features is feasible. Although functional differences with adipose-derived stem cells were limited, based on their gene expression, pericard- and pleura-derived stem cells may represent a superior autologous cell source for cell seeding in tracheal tissue engineering.


Asunto(s)
Células Madre Multipotentes/citología , Pericardio/citología , Pleura/citología , Tráquea/citología , Adipocitos/citología , Adipogénesis/fisiología , Tejido Adiposo/citología , Animales , Células de la Médula Ósea/citología , Diferenciación Celular/fisiología , Condrogénesis/fisiología , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/fisiología , Células Madre/citología , Porcinos , Ingeniería de Tejidos/métodos
10.
Indian J Pathol Microbiol ; 64(4): 683-686, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34673586

RESUMEN

BACKGROUND: The "Ring-and-Sling" complex (RSC) comprises congenital tracheal stenosis and an abnormal origin/course of the left pulmonary artery. Based on clinical and imaging studies, the luminal narrowing is assumed to be as a result of rings cartilage (forming an "O"). AIMS: This is a postmortem based study of tracheal histology in infants after an autopsy encounter of a case of RSC. SUBJECT AND METHODS: RSC was identified in an infant at autopsy. The tracheal histomorphology revealed the presence of cartilaginous plates (instead of rings) and fibro-elastotic proliferation at the site of trachealis muscle. These changes prompted a study on variations in the histology of the trachea (with no known anomaly) in 35 autopsied neonates and infants. The transverse sections of the trachea were taken at one or more levels (Level 1 - at the level of the thyroid, Level 2 - midway between the thyroid and the carina, and Level 3 - just above the carina. STATISTICAL ANALYSIS: Epi-info software (v1.4.3, CD, US). RESULTS: On histology, 83 sections showed the trachealis muscle on the posterior aspect. A single semicircular cartilage was identified in only 17 of the 83 sections studied (20.5%, 6 in level 1, 9 in level 2 and 2 in level 3). In the remaining 66 sections (79.5%), the cartilage was disposed as multiple plates, ranging in number from 2 to 10. No significant association was found between semicircular cartilage rings and age, sex, gestational age, and level of section (P < 0.05). However, 14 cases with sectioning at all three levels were taken into account; all levels showed more cartilaginous plates compared to single rings, which were more common at level 1 (P > 0.05). CONCLUSIONS: The "ring" in RSC and normal infantile tracheas show cartilage plates with intermittent semicircular cartilage rings. These findings may have surgical implications for tracheal anomalies and bode favorable surgical outcomes.


Asunto(s)
Anomalías Congénitas/diagnóstico , Anomalías Congénitas/fisiopatología , Tráquea/citología , Tráquea/fisiopatología , Estenosis Traqueal/congénito , Estenosis Traqueal/diagnóstico , Estenosis Traqueal/fisiopatología , Autopsia/estadística & datos numéricos , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino
11.
Int J Mol Sci ; 22(19)2021 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-34638663

RESUMEN

Decellularization has emerged as a potential solution for tracheal replacement. As a fully decellularized graft failed to achieve its purposes, the de-epithelialization partial decellularization protocol appeared to be a promising approach for fabricating scaffolds with preserved mechanical properties and few immune rejection responses after transplantation. Nevertheless, a lack of appropriate concurrent epithelialization treatment can lead to luminal stenosis of the transplant and impede its eventual success. To improve re-epithelialization, autologous nasal epithelial cell sheets generated by our cell sheet engineering platform were utilized in this study under an in vivo rabbit model. The newly created cell sheets have an intact and transplantable appearance, with their specific characteristics of airway epithelial origin being highly expressed upon histological and immunohistochemical analysis. Subsequently, those cell sheets were incorporated with a partially decellularized tracheal graft for autograft transplantation under tracheal partial resection models. The preliminary results two months post operation demonstrated that the transplanted patches appeared to be wholly integrated into the host trachea with adequate healing of the luminal surface, which was confirmed via endoscopic and histologic evaluations. The satisfactory result of this hybrid scaffold protocol could serve as a potential solution for tracheal reconstructions in the future.


Asunto(s)
Células Epiteliales/citología , Nariz/citología , Tráquea/citología , Animales , Estudios Transversales , Matriz Extracelular/fisiología , Masculino , Conejos , Procedimientos de Cirugía Plástica/métodos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Trasplante Autólogo/métodos
12.
Int J Mol Sci ; 22(19)2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34638978

RESUMEN

Natural or experimental infection of domestic cats and virus transmission from humans to captive predatory cats suggest that felids are highly susceptible to SARS-CoV-2 infection. However, it is unclear which cells and compartments of the respiratory tract are infected. To address this question, primary cell cultures derived from the nose, trachea, and lungs of cat and lion were inoculated with SARS-CoV-2. Strong viral replication was observed for nasal mucosa explants and tracheal air-liquid interface cultures, whereas replication in lung slices was less efficient. Infection was mainly restricted to epithelial cells and did not cause major pathological changes. Detection of high ACE2 levels in the nose and trachea but not lung further suggests that susceptibility of feline tissues to SARS-CoV-2 correlates with ACE2 expression. Collectively, this study demonstrates that SARS-CoV-2 can efficiently replicate in the feline upper respiratory tract ex vivo and thus highlights the risk of SARS-CoV-2 spillover from humans to felids.


Asunto(s)
COVID-19/veterinaria , Gatos/virología , Leones/virología , Enzima Convertidora de Angiotensina 2/análisis , Animales , COVID-19/transmisión , COVID-19/virología , Enfermedades de los Gatos/transmisión , Enfermedades de los Gatos/virología , Células Cultivadas , Susceptibilidad a Enfermedades , Humanos , Pulmón/citología , Pulmón/virología , Nariz/citología , Nariz/virología , SARS-CoV-2/aislamiento & purificación , Tráquea/citología , Tráquea/virología
13.
Nat Protoc ; 16(12): 5484-5532, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34716451

RESUMEN

The development of biocompatible and precisely printable bioink addresses the growing demand for three-dimensional (3D) bioprinting applications in the field of tissue engineering. We developed a methacrylated photocurable silk fibroin (SF) bioink for digital light processing 3D bioprinting to generate structures with high mechanical stability and biocompatibility for tissue engineering applications. Procedure 1 describes the synthesis of photocurable methacrylated SF bioink, which takes 2 weeks to complete. Digital light processing is used to fabricate 3D hydrogels using the bioink (1.5 h), which are characterized in terms of methacrylation, printability, mechanical and rheological properties, and biocompatibility. The physicochemical properties of the bioink can be modulated by varying photopolymerization conditions such as the degree of methacrylation, light intensity, and concentration of the photoinitiator and bioink. The versatile bioink can be used broadly in a range of applications, including nerve tissue engineering through co-polymerization of the bioink with graphene oxide, and for wound healing as a sealant. Procedure 2 outlines how to apply 3D-printed SF hydrogels embedded with chondrocytes and turbinate-derived mesenchymal stem cells in one specific in vivo application, trachea tissue engineering, which takes 2-9 weeks.


Asunto(s)
Bioimpresión/métodos , Fibroínas/química , Hidrogeles/química , Tejido Nervioso/efectos de los fármacos , Ingeniería de Tejidos/métodos , Tráquea/efectos de los fármacos , Animales , Condrocitos/citología , Condrocitos/efectos de los fármacos , Condrocitos/fisiología , Fibroínas/farmacología , Grafito/química , Humanos , Hidrogeles/farmacología , Luz , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/fisiología , Metacrilatos/química , Ratones , Tejido Nervioso/citología , Tejido Nervioso/fisiología , Impresión Tridimensional/instrumentación , Conejos , Andamios del Tejido , Tráquea/citología , Tráquea/fisiología , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología
14.
Biol Open ; 10(9)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34396394

RESUMEN

The tracheal basal cells (BCs) function as stem cells to maintain the epithelium in steady state and repair it after injury. The airway is surrounded by cartilage ventrolaterally and smooth muscle dorsally. Lineage tracing using Krt5-CreER shows dorsal BCs produce more, larger, clones than ventral BCs. Large clones were found between cartilage and smooth muscle where subpopulation of dorsal BCs exists. Three-dimensional organoid culture of BCs demonstrated that dorsal BCs show higher colony forming efficacy to ventral BCs. Gene ontology analysis revealed that genes expressed in dorsal BCs are enriched in wound healing while ventral BCs are enriched in response to external stimulus and immune response. Significantly, ventral BCs express Myostatin, which inhibits the growth of smooth muscle cells, and HGF, which facilitates cartilage repair. The results support the hypothesis that BCs from the dorso-ventral airways have intrinsic molecular and behavioural differences relevant to their in vivo function.


Asunto(s)
Diferenciación Celular , Células Epiteliales/fisiología , Heterogeneidad Genética , Células Madre/citología , Tráquea/citología , Ontología de Genes , Humanos
15.
Respir Res ; 22(1): 238, 2021 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-34446024

RESUMEN

BACKGROUND: Childhood asthma is a common respiratory disease characterized by airway inflammation. Tumor necrosis factor-α-induced protein 8-like 2 (TIPE2) has been found to be involved in the progression of asthma. This study aimed to explore the role of TIPE2 in the regulation of airway smooth muscle cells (ASMCs), which are one of the main effector cells in the development of asthma. MATERIALS AND METHODS: ASMCs were transfected with pcDNA3.0-TIPE2 or si-TIPE2 for 48 h and then treated with platelet-derived growth factor (PDGF)-BB. Cell proliferation of ASMCs was measured using the MTT assay. Cell migration of ASMCs was determined by a transwell assay. The mRNA expression levels of calponin and smooth muscle protein 22α (SM22α) were measured using qRT-PCR. The levels of TIPE2, calponin, SM22α, PI3K, p-PI3K, Akt, and p-Akt were detected by Western blotting. RESULTS: Our results showed that PDGF-BB treatment significantly reduced TIPE2 expression at both the mRNA and protein levels in ASMCs. Overexpression of TIPE2 inhibited PDGF-BB-induced ASMC proliferation and migration. In addition, overexpression of TIPE2 increased the expression of calponin and SM22α in PDGF-BB-stimulated ASMCs. However, an opposite effect was observed with TIPE2 knockdown. Furthermore, TIPE2 overexpression blocked PDGF-BB-induced phosphorylation of PI3K and Akt, whereas the expression of p-PI3K and p-Akt were aggravated by TIPE2 knockdown. Additionally, the effects of TIPE2 overexpression and TIPE2 knockdown were altered by IGF-1 and LY294002 treatments, respectively. CONCLUSIONS: Our findings demonstrate that TIPE2 inhibits PDGF-BB-induced ASMC proliferation, migration, and phenotype switching via the PI3K/Akt signaling pathway. Thus, TIPE2 may be a potential therapeutic target for the treatment of asthma.


Asunto(s)
Becaplermina/toxicidad , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Músculo Liso/metabolismo , Miocitos del Músculo Liso/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ratones , Ratones Endogámicos C57BL , Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Fenotipo , Inhibidores de las Quinasa Fosfoinosítidos-3/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Tráquea/citología , Tráquea/efectos de los fármacos , Tráquea/metabolismo
16.
STAR Protoc ; 2(3): 100683, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34355203

RESUMEN

Airway basal cells play an essential role in the maintenance of the airway epithelium. Here, we provide a detailed directed differentiation protocol to generate ''induced basal cells (iBCs)'' from human pluripotent stem cells. iBCs recapitulate biological and functional properties of airway basal cells including mucociliary differentiation in vitro or in vivo in tracheal xenografts, facilitating the study of inherited and acquired diseases of the airway, as well as potential use in regenerative medicine. For complete details on the use and execution of this protocol, please refer to Hawkins et al. (2021).


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Sistema Respiratorio/citología , Ingeniería de Tejidos/métodos , Diferenciación Celular/fisiología , Células Cultivadas , Endodermo/citología , Células Epiteliales/citología , Epitelio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Pulmón/citología , Organoides/citología , Células Madre Pluripotentes/citología , Tráquea/citología
17.
Physiol Rep ; 9(15): e14928, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34382377

RESUMEN

The cystic fibrosis transmembrane conductance regulator (CFTR) is an anion channel whose dysfunction causes cystic fibrosis (CF). The loss of CFTR function in pulmonary epithelial cells causes surface dehydration, mucus build-up, inflammation, and bacterial infections that lead to lung failure. Little has been done to evaluate the effects of lipid perturbation on CFTR activity, despite CFTR residing in the plasma membrane. This work focuses on the acute effects of sphingomyelinase (SMase), a bacterial virulence factor secreted by CF relevant airway bacteria which degrades sphingomyelin into ceramide and phosphocholine, on the electrical circuitry of pulmonary epithelial monolayers. We report that basolateral SMase decreases CFTR-mediated transepithelial anion secretion in both primary bronchial and tracheal epithelial cells from explant tissue, with current CFTR modulators unable to rescue this effect. Focusing on primary cells, we took a holistic ion homeostasis approach to determine a cause for reduced anion secretion following SMase treatment. Using impedance analysis, we determined that basolateral SMase inhibits apical and basolateral conductance in non-CF primary cells without affecting paracellular permeability. In CF primary airway cells, correction with clinically relevant CFTR modulators did not prevent SMase-mediated inhibition of CFTR currents. Furthermore, SMase was found to inhibit only apical conductance in these cells. Future work should determine the mechanism for SMase-mediated inhibition of CFTR currents, and further explore the clinical relevance of SMase and sphingolipid imbalances.


Asunto(s)
Aniones/metabolismo , Bronquios/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Staphylococcus aureus/enzimología , Tráquea/metabolismo , Bronquios/citología , Polaridad Celular , Células Cultivadas , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Células Epiteliales/citología , Humanos , Transporte Iónico , Mutación , Esfingomielina Fosfodiesterasa/genética , Tráquea/citología
18.
Artif Organs ; 45(12): 1491-1500, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34310703

RESUMEN

In tracheal replacement transplantation, prelamination is a critical stage. Nowadays, the most widely used prelamination technique is the prethoracic fascia flap with lateral thoracic artery. We propose a flap based on the internal thoracic artery, which allows a relatively non-aggressive double organ implant, and we have tested its efficacy in decellularized tracheas. Tracheas of albino New Zealand rabbits were decellularized following a protocol that uses detergents and cryogenization, sterilized with 1kGy gamma radiation, and tutorized with a stent. Bilateral pedicled flaps made of pectoral fascia and a muscular component were harvested through a longitudinal 3-cm central thoracic incision, wrapping the tracheas with them in 16 rabbits, remaining them implanted for 2, 4, 8, and 12 weeks. The tracheas were then studied histologically using standard stainings plus immunohistochemistry (CD31). The models were adjusted with Bayesian statistics using ordinal regression; results as odds ratios and credibility intervals. All analysis were performed using R software. Acute inflammatory cell invasion was observed at 2 weeks, which almost disappeared at week 8 after implant. Only macrophages and giant cells increased between Weeks 8 and 12 (OR 10.487, CI [1.603-97.327]). The cartilage maintained its structure, with slight signs of ischemia in a few cases. New CD31-positive vessels were observed from Week 2 and increasing thereafter, reaching a maximum peak at Week 8. We propose a bilateral implant technique that is viable and effective as a prelamination option for two concurrent tracheas, achieving perfect vascularization and integration of the organ with hardly any inflammatory response in the medium or long term.


Asunto(s)
Bioprótesis , Ingeniería de Tejidos/métodos , Tráquea/trasplante , Animales , Sistema Libre de Células , Masculino , Conejos , Tráquea/citología , Trasplante Homólogo/métodos
19.
Vet Microbiol ; 259: 109129, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34087675

RESUMEN

Caprine parainfluenza virus type 3 (CPIV3) is one of the most important viral respiratory pathogens of goat. Accumulating evidence demonstrates that apoptosis is a cellular mechanism for the host response to pathogens, and it participates in regulating viral replication. However, there is little study on CPIV3-induced host cells apoptosis. In this study, primary goat tracheal epithelial (GTE) cells were established as a cellular model that is permissive to CPIV3 infection. Then, we showed that CPIV3 infection induced apoptosis in GTE cells, as determined by morphological changes, flow cytometry and TUNEL assay. Moreover, Caspase activity and the expression of pro-apoptotic genes further suggested that CPIV3 induced apoptosis by activating both the intrinsic and extrinsic pathways. Mechanistically, the ability of CPIV3 to induce apoptosis was activated by N protein, and the viral protein increased CPIV3 replication through effecting apoptosis. Overall, our findings showed that GTE cells that will enable further analysis of CPIV3 infection and offers novel insights into the mechanisms of CPIV3-induced apoptosis in host cells.


Asunto(s)
Apoptosis/genética , Proteínas de la Nucleocápside/genética , Virus de la Parainfluenza 3 Humana/química , Virus de la Parainfluenza 3 Humana/genética , Infecciones por Respirovirus/genética , Infecciones por Respirovirus/veterinaria , Replicación Viral/genética , Animales , Células Cultivadas , Células Epiteliales/virología , Enfermedades de las Cabras/virología , Cabras/virología , Proteínas de la Nucleocápside/metabolismo , Virus de la Parainfluenza 3 Humana/patogenicidad , Infecciones por Respirovirus/virología , Tráquea/citología
20.
FASEB J ; 35(7): e21674, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34115899

RESUMEN

Current therapeutic approaches to avoid or reverse bronchoconstriction rely primarily on ß2 adrenoceptor agonists (ß-agonists) that regulate pharmacomechanical coupling/cross bridge cycling in airway smooth muscle (ASM). Targeting actin cytoskeleton polymerization in ASM represents an alternative means to regulate ASM contraction. Herein we report the cooperative effects of targeting these distinct pathways with ß-agonists and inhibitors of the mammalian Abelson tyrosine kinase (Abl1 or c-Abl). The cooperative effect of ß-agonists (isoproterenol) and c-Abl inhibitors (GNF-5, or imatinib) on contractile agonist (methacholine, or histamine) -induced ASM contraction was assessed in cultured human ASM cells (using Fourier Transfer Traction Microscopy), in murine precision cut lung slices, and in vivo (flexiVent in mice). Regulation of intracellular signaling that regulates contraction (pMLC20, pMYPT1, pHSP20), and actin polymerization state (F:G actin ratio) were assessed in cultured primary human ASM cells. In each (cell, tissue, in vivo) model, c-Abl inhibitors and ß-agonist exhibited additive effects in either preventing or reversing ASM contraction. Treatment of contracted ASM cells with c-Abl inhibitors and ß-agonist cooperatively increased actin disassembly as evidenced by a significant reduction in the F:G actin ratio. Mechanistic studies indicated that the inhibition of pharmacomechanical coupling by ß-agonists is near optimal and is not increased by c-Abl inhibitors, and the cooperative effect on ASM relaxation resides in further relaxation of ASM tension development caused by actin cytoskeleton depolymerization, which is regulated by both ß-agonists and c-Abl inhibitors. Thus, targeting actin cytoskeleton polymerization represents an untapped therapeutic reserve for managing airway resistance.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Sinergismo Farmacológico , Contracción Muscular , Relajación Muscular , Músculo Liso/fisiología , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Tráquea/fisiología , Citoesqueleto de Actina/metabolismo , Animales , Antineoplásicos/farmacología , Benzamidas/farmacología , Humanos , Mesilato de Imatinib/farmacología , Isoproterenol/farmacología , Ratones , Ratones Endogámicos C57BL , Músculo Liso/citología , Músculo Liso/efectos de los fármacos , Pirimidinas/farmacología , Transducción de Señal , Tráquea/citología , Tráquea/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA